1
|
Schmid A, Lausch U, Runkel A, Kiefer J, Pauli T, Boerries M, Bogner B, Eisenhardt SU, Braig D. Improved Quantification of Circulating Tumor DNA in Translocation-Associated Myxoid Liposarcoma by Simultaneous Detection of Breakpoints and Single Nucleotide Variants. Cancer Med 2025; 14:e70704. [PMID: 39980272 PMCID: PMC11842865 DOI: 10.1002/cam4.70704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 11/19/2024] [Accepted: 02/08/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Myxoid liposarcomas (MLS) can exhibit a disseminated metastatic pattern, necessitating extensive diagnostics during follow-up. With no tumor markers available, early diagnosis of recurrences and tumor monitoring is difficult. The detection of circulating tumor DNA (ctDNA; liquid biopsy) in MLS with the characteristic translocations t(12;16) and t(12;22) can provide an additional diagnostic. However, due to the often very low tumor fraction, distinguishing actual tumor variants from sequencing artifacts remains a key challenge. METHODS Using MLS as a model for translocation-driven tumors, this study evaluates a refined analytical approach for detecting both single nucleotide variants (SNVs) and structural variants (SVs) with the highest possible sensitivity and specificity. Different analysis pipelines using Unique Molecular Identifiers (UMIs) were compared in dilution series of tumor DNA from MLS patients (n = 11) and a cell line. The results were validated on plasma samples (n = 36) from two MLS patients and one patient with Synovial Sarcoma (SS). RESULTS In dilution series, the use of UMIs significantly reduced false positive events in SNV analysis while maintaining high sensitivity without significant differences. In SV analysis, the effect of UMIs was not consistently detectable, as some dilution series exhibited no false positive events even without UMI correction. Additional filter criteria further improved specificity without significantly compromising assay sensitivity. Validation on patient plasma samples confirmed these findings, demonstrating the advantages of the differentiated analytical approach. CONCLUSION By integrating a refined analytical approach for SNVs and SVs, we achieved reliable ctDNA detection that corresponded with the clinical course of the patients' disease. This method enables non-invasive tumor detection in translocation-driven tumors with low mutational burden and can easily be adapted into routine clinical diagnostics.
Collapse
Affiliation(s)
- A. Schmid
- Department of Plastic and Hand SurgeryMedical Center ‐ University of Freiburg, Faculty of Medicine, University of FreiburgFreiburgGermany
| | - U. Lausch
- Department of Plastic and Hand SurgeryMedical Center ‐ University of Freiburg, Faculty of Medicine, University of FreiburgFreiburgGermany
| | - A. Runkel
- Department of Plastic and Hand SurgeryMedical Center ‐ University of Freiburg, Faculty of Medicine, University of FreiburgFreiburgGermany
| | - J. Kiefer
- Department of Plastic and Hand SurgeryMedical Center ‐ University of Freiburg, Faculty of Medicine, University of FreiburgFreiburgGermany
| | - T. Pauli
- Institute of Medical Bioinformatics and Systems MedicineMedical Center–University of Freiburg, Faculty of Medicine, University of FreiburgFreiburgGermany
| | - M. Boerries
- Institute of Medical Bioinformatics and Systems MedicineMedical Center–University of Freiburg, Faculty of Medicine, University of FreiburgFreiburgGermany
| | - B. Bogner
- Department of RadiologyMedical Center–University of Freiburg, Faculty of Medicine, University of FreiburgFreiburgGermany
| | - S. U. Eisenhardt
- Department of Plastic and Hand SurgeryMedical Center ‐ University of Freiburg, Faculty of Medicine, University of FreiburgFreiburgGermany
| | - D. Braig
- Department of Plastic and Hand SurgeryMedical Center ‐ University of Freiburg, Faculty of Medicine, University of FreiburgFreiburgGermany
| |
Collapse
|
2
|
Abu-Hijlih R, Sharaf B, Salah S, Bani Hani H, Alqaisieh M, Alzibdeh A, Ababneh L, Mahafdah S, Abdel-Razeq H. Germline Genetic Mutations in Adult Patients with Sarcoma: Insight into the Middle East Genetic Landscape. Cancers (Basel) 2024; 16:1668. [PMID: 38730621 PMCID: PMC11083501 DOI: 10.3390/cancers16091668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/21/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
Data on germline mutations in soft tissue and bone sarcomas are scarce. We sought to identify the prevalence of germline mutations in adult sarcoma patients treated at a tertiary cancer center. Newly diagnosed patients were offered germline genetic testing via an 84-gene panel. The prevalence of pathogenic germline variants (PGVs) and their association with disease-, and patient- related factors are reported. A total of 87 patients were enrolled, the median age was 48 (19-78) years, and 47 (54%) were females. Gastrointestinal stromal tumors (n = 12, 13.8%), liposarcoma (n = 10, 11.5%), and Ewing sarcoma (n = 10, 11.5%) were the main subtypes. A total of 20 PGVs were detected in 18 (20.7%) patients. Variants of uncertain significance, in the absence of PGVs, were detected in 40 (45.9%) patients. Young age (p = 0.031), presence of a second primary cancer (p = 0.019), and female gender (p = 0.042) were correlated with the presence of PGVs. All identified PGVs have potential clinical actionability and cascade testing, and eight (44.44%) suggested eligibility for a targeted therapy. Almost one in five adult patients with soft tissue and bone sarcomas harbor pathogenic or likely pathogenic variants. Many of these variants are potentially actionable, and almost all have implications on cancer screening and family counselling. In this cohort from the Middle East, younger age, presence of a second primary tumor, and female gender were significantly associated with higher PGVs rates. Larger studies able to correlate treatment outcomes with genetic variants are highly needed.
Collapse
Affiliation(s)
- Ramiz Abu-Hijlih
- Department of Radiation Oncology, King Hussein Cancer Center, Amman 11941, Jordan; (R.A.-H.); (A.A.)
| | - Baha Sharaf
- Department of Internal Medicine, King Hussein Cancer Center, Amman 11941, Jordan; (B.S.); (S.S.); (H.B.H.); (M.A.)
| | - Samer Salah
- Department of Internal Medicine, King Hussein Cancer Center, Amman 11941, Jordan; (B.S.); (S.S.); (H.B.H.); (M.A.)
| | - Hira Bani Hani
- Department of Internal Medicine, King Hussein Cancer Center, Amman 11941, Jordan; (B.S.); (S.S.); (H.B.H.); (M.A.)
| | - Mohammad Alqaisieh
- Department of Internal Medicine, King Hussein Cancer Center, Amman 11941, Jordan; (B.S.); (S.S.); (H.B.H.); (M.A.)
| | - Abdulla Alzibdeh
- Department of Radiation Oncology, King Hussein Cancer Center, Amman 11941, Jordan; (R.A.-H.); (A.A.)
| | - Layan Ababneh
- School of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan;
| | - Suleiman Mahafdah
- Department of Surgery, Royal Jordanian Medical Services, Amman 11855, Jordan;
| | - Hikmat Abdel-Razeq
- Department of Internal Medicine, King Hussein Cancer Center, Amman 11941, Jordan; (B.S.); (S.S.); (H.B.H.); (M.A.)
- School of Medicine, The University of Jordan, Amman 11942, Jordan
| |
Collapse
|
3
|
Huang X, Li G, Li L, Wang J, Shen J, Chen Y, Yu W, Chen A, Wu T, Ma J, Ling B, He L, Chen X. Establishing an RNA fusions panel in soft tissue sarcoma with clinical validation. Sci Rep 2023; 13:4403. [PMID: 36928336 PMCID: PMC10020547 DOI: 10.1038/s41598-023-29511-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 02/06/2023] [Indexed: 03/18/2023] Open
Abstract
The diagnosis and classification of soft tissue sarcomas (STS) remain challenging because of the rarity and overlapping morphologic manifestations of diverse STS subtypes. Characteristic gene fusions are commonly detected in STS and represent useful diagnostic markers. This study established and validated a custom-designed RNA sequencing panel that identified 64 gene fusions in STS. The analytical performance validation yielded excellent accuracy, with 100% (95% CI, 94.40%-100%) sensitivity and 93.33% (95% CI, 68.05%-99.83%) specificity. Clinical performances were further confirmed with 145 clinical formalin-fixed and paraffin-embedded (FFPE) samples from STS patients. Fusions were detected in 40% of samples (58/145). The common fusions SS18-SSX family, EWSR1-related fusions, COL1A1-PDGFB, FOXO1-associated fusions, and FUS-associated fusions were identified in corresponding STS subtypes. The RNA panel detected specific fusions in several cases where no conclusive diagnosis can be made based on the morphology and immunohistochemistry results. Data collected in this study demonstrate that the RNA fusions panel can better classify STS subtypes and serve as a good supplement for histopathology, exhibiting a great potential for the STS precise diagnosis.
Collapse
Affiliation(s)
- Xiaoqiang Huang
- Guangzhou KingMed Center for Clinical Laboratory Co. Ltd., Guangzhou, China
- Guangzhou KingMed Diagnostics Group Co. Ltd., Guangzhou, China
| | - Guibin Li
- Guangzhou KingMed Transformative Medicine Institute Co. Ltd., Guangzhou, China
| | - Linghua Li
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jian Wang
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jianru Shen
- Guangzhou KingMed Diagnostics Group Co. Ltd., Guangzhou, China.
| | - Yao Chen
- Guangzhou KingMed Diagnostics Group Co. Ltd., Guangzhou, China
| | - Wuzhong Yu
- Guangzhou KingMed Diagnostics Group Co. Ltd., Guangzhou, China
| | - Ailin Chen
- Guangzhou KingMed Diagnostics Group Co. Ltd., Guangzhou, China
| | - Tao Wu
- Guangzhou KingMed Center for Clinical Laboratory Co. Ltd., Guangzhou, China
| | - Ji Ma
- Guangzhou KingMed Diagnostics Group Co. Ltd., Guangzhou, China
| | - Bao Ling
- Shenzhen KingMed Medical Laboratory, Shenzhen, China
| | - Liang He
- Shenzhen KingMed Medical Laboratory, Shenzhen, China
| | - Xudan Chen
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
4
|
Vargas AC, Heyer EE, Cheah AL, Bonar F, Jones M, Maclean FM, Gill AJ, Blackburn J. Improving sarcoma classification by using RNA hybridisation capture sequencing in sarcomas of uncertain histogenesis of young individuals. Pathology 2023; 55:478-485. [PMID: 36906400 DOI: 10.1016/j.pathol.2022.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 10/14/2022] [Accepted: 11/22/2022] [Indexed: 02/16/2023]
Abstract
Our aim was to utilise a 241-gene RNA hybridisation capture sequencing (CaptureSeq) gene panel to identify unexpected fusions in undifferentiated, unclassified or partly classified sarcomas of young individuals (<40 years). The purpose was to determine the utility and yield of a large, targeted fusion panel as a tool for classifying tumours that do not fit typical diagnostic entities at the time of the original diagnosis. RNA hybridisation capture sequencing was performed on 21 archival resection specimens. Successful sequencing was obtained in 12 of 21 samples (57%), two of which (16.6%) harboured translocations. A novel NEAT1::GLI1 fusion, not previously reported in the literature, presented in a young patient with a tumour in the retroperitoneum, which displayed low grade epithelioid cells. The second case, a localised lung metastasis in a young male, demonstrated a EWSR1::NFATC2 translocation. No targeted fusions were identified in the remaining 83.4% (n=10) of cases. Forty-three per cent of the samples failed sequencing as a result of RNA degradation. RNA-based sequencing is an important tool, which helps to redefine the classification of unclassified or partly classified sarcomas of young adults by identifying pathogenic gene fusions in up to 16.6% of the cases. Unfortunately, 43% of the samples underwent significant RNA degradation, falling below the sequencing threshold. As CaptureSeq is not yet available in routine pathology practice, increasing awareness of the yield, failure rate and possible aetiological factors for RNA degradation is fundamental to maximise laboratory procedures to improve RNA integrity, allowing the potential identification of significant gene alterations in solid tumours.
Collapse
Affiliation(s)
- Ana Cristina Vargas
- Department of Anatomical Pathology, Sonic Healthcare-Douglass Hanly Moir Pathology, Macquarie Park, NSW, Australia; Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia; Sydney Medical School, University of Sydney, Sydney, NSW, Australia.
| | - Erin E Heyer
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, NSW, Australia; St Vincent's Clinical School, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Alison L Cheah
- Department of Anatomical Pathology, Sonic Healthcare-Douglass Hanly Moir Pathology, Macquarie Park, NSW, Australia
| | - Fiona Bonar
- Department of Anatomical Pathology, Sonic Healthcare-Douglass Hanly Moir Pathology, Macquarie Park, NSW, Australia
| | - Martin Jones
- Department of Anatomical Pathology, Sonic Healthcare-Douglass Hanly Moir Pathology, Macquarie Park, NSW, Australia
| | - Fiona M Maclean
- Department of Anatomical Pathology, Sonic Healthcare-Douglass Hanly Moir Pathology, Macquarie Park, NSW, Australia; Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia; Department of Clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University, Macquarie Park, NSW, Australia
| | - Anthony J Gill
- Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia; Sydney Medical School, University of Sydney, Sydney, NSW, Australia; Department of Anatomical Pathology, NSW Health Pathology, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - James Blackburn
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, NSW, Australia; St Vincent's Clinical School, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
5
|
Lopes-Brás R, Lopez-Presa D, Esperança-Martins M, Melo-Alvim C, Gallego L, Costa L, Fernandes I. Genomic Profiling of Sarcomas: A Promising Weapon in the Therapeutic Arsenal. Int J Mol Sci 2022; 23:14227. [PMID: 36430703 PMCID: PMC9693140 DOI: 10.3390/ijms232214227] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/12/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022] Open
Abstract
Sarcomas are rare malignant mesenchymal neoplasms, and the knowledge of tumor biology and genomics is scarce. Chemotherapy is the standard of care in advanced disease, with poor outcomes. Identifying actionable genomic alterations may offer effective salvage therapeutic options when previous lines have failed. Here, we report a retrospective cohort study of sarcoma patients followed at our center and submitted to comprehensive genomic profiling between January 2020 and June 2021. Thirty patients were included, most (96.7%) with reportable genomic alterations. The most common alterations were linked to cell cycle regulation (TP53, CDKN2A/B, and RB1 deletions and CDK4, MDM2, and MYC amplifications). Most patients (96.7%) had microsatellite stability and low tumor mutational burden (≤10 muts/megabase (Mb); median 2 Muts/Mb). Two-thirds of patients had actionable mutations for targeted treatments, including five cases with alterations amenable to targeted therapies with clinical benefit within the patient's tumor type, ten cases with targetable alterations with clinical benefit in other tumor types, and five cases with alterations amenable to targeting with drugs under investigation in a clinical trial setting. A significant proportion of cases in this study had actionable genomic alterations with available targeted drugs. Next-generation sequencing is a feasible option for identifying molecular drivers that can provide therapeutic options for individual patients. Molecular Tumor Boards should be implemented in the clinical practice to discuss genomic findings and inform clinically relevant targeted therapies.
Collapse
Affiliation(s)
- Raquel Lopes-Brás
- Department of Medical Oncology, Hospital Santa Maria, Centro Hospitalar Universitário Lisboa Norte, 1649-028 Lisbon, Portugal
| | - Dolores Lopez-Presa
- Department of Pathology, Hospital Santa Maria, Centro Hospitalar Universitário Lisboa Norte, 1649-028 Lisbon, Portugal
| | - Miguel Esperança-Martins
- Department of Medical Oncology, Hospital Santa Maria, Centro Hospitalar Universitário Lisboa Norte, 1649-028 Lisbon, Portugal
- Sérgio Dias Lab, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal
- Luís Costa Lab, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Cecília Melo-Alvim
- Department of Medical Oncology, Hospital Santa Maria, Centro Hospitalar Universitário Lisboa Norte, 1649-028 Lisbon, Portugal
| | - Lina Gallego
- Luís Costa Lab, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Luís Costa
- Department of Medical Oncology, Hospital Santa Maria, Centro Hospitalar Universitário Lisboa Norte, 1649-028 Lisbon, Portugal
- Luís Costa Lab, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Isabel Fernandes
- Department of Medical Oncology, Hospital Santa Maria, Centro Hospitalar Universitário Lisboa Norte, 1649-028 Lisbon, Portugal
- Luís Costa Lab, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal
| |
Collapse
|
6
|
Reimers MA, Sehn JK, Van Tine BA, Smith ZL. Primary Prostatic Synovial Sarcoma With Pulmonary Metastases Identified by Routine Next-Generation Sequencing. JCO Precis Oncol 2022; 5:1133-1140. [PMID: 34994631 DOI: 10.1200/po.21.00051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Melissa A Reimers
- Division of Oncology, Department of Internal Medicine, Section of Medical Oncology, Washington University in St Louis, St Louis, MO.,Siteman Cancer Center, Washington University in St Louis, St Louis, MO
| | - Jennifer K Sehn
- Department of Pathology and Immunology, Washington University in St Louis, St Louis, MO
| | - Brian A Van Tine
- Division of Oncology, Department of Internal Medicine, Section of Medical Oncology, Washington University in St Louis, St Louis, MO.,Siteman Cancer Center, Washington University in St Louis, St Louis, MO.,Department of Pediatric Hematology and Oncology, St Louis Children's Hospital, St Louis, MO
| | - Zachary L Smith
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO.,Division of Urologic Surgery, Department of Surgery, Washington University in St Louis, St Louis, MO
| |
Collapse
|
7
|
Gaiser T, Sauer C, Marx A, Jakob J, Kasper B, Hohenberger P, Hirsch D, Ronellenfitsch U. Molecular and Pathological Profiling of Corresponding Treatment-Naïve and Neoadjuvant Pazopanib-Treated High-Risk Soft Tissue Sarcoma Samples of the GISG-04/NOPASS Study. BIOLOGY 2021; 10:biology10070639. [PMID: 34356494 PMCID: PMC8301157 DOI: 10.3390/biology10070639] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 07/07/2021] [Indexed: 01/25/2023]
Abstract
In the framework of the German Interdisciplinary Sarcoma Group GISG-04/NOPASS trial, we evaluated soft tissue sarcoma samples taken before and after neoadjuvant pazopanib therapy using histopathology and next generation sequencing (NGS) to find potential predictive biomarkers. We also aimed to improve the genetically based sarcoma classification and to elucidate additional potentially druggable mutations. In total, 30 tumor samples from 18 patients consisting of 12 pre-therapeutic biopsies and 18 resection specimens following neoadjuvant pazopanib therapy were available for analyses. NGS was performed with the Oncomine Focus Assay (Ion Torrent) covering 0.03 Mb of DNA and enabled the detection of genetic variants in 52 cancer-relevant genes. Pathological analysis showed significant regression (≥50%) after pazopanib treatment in only one undifferentiated (pleomorphic) sarcoma. NGS analyses revealed a very high frequency of CDK4 amplification (88%; 7/8) in the group of dedifferentiated liposarcoma. In addition, two potentially druggable mutations, a MAP2K1 missense mutation (E203K) and a BRAF missense mutation (V600E), were traceable in two undifferentiated (pleomorphic) sarcoma patients (11%; 2/18). Our findings demonstrate that NGS testing is a powerful technology helping to improve diagnostic accuracy and offering some patients the chance for personalized medicine even in a "mutation unlikely" cohort like STS.
Collapse
Affiliation(s)
- Timo Gaiser
- Institute of Pathology, University Medical Centre Mannheim, University of Heidelberg, 68305 Mannheim, Germany; (C.S.); (A.M.); (D.H.)
- Correspondence: ; Tel.: +49-621-383-2876; Fax: +49-621-383-2005
| | - Christian Sauer
- Institute of Pathology, University Medical Centre Mannheim, University of Heidelberg, 68305 Mannheim, Germany; (C.S.); (A.M.); (D.H.)
| | - Alexander Marx
- Institute of Pathology, University Medical Centre Mannheim, University of Heidelberg, 68305 Mannheim, Germany; (C.S.); (A.M.); (D.H.)
| | - Jens Jakob
- Department of General, Visceral and Child Surgery, University Medical Center Göttingen, 37075 Göttingen, Germany;
| | - Bernd Kasper
- Sarcoma Unit, Interdisciplinary Tumor Center Mannheim, Mannheim University Medical Center, 68305 Mannheim, Germany;
| | - Peter Hohenberger
- Division of Surgical Oncology and Thoracic Surgery, University Medical Center Mannheim, 68167 Mannheim, Germany;
| | - Daniela Hirsch
- Institute of Pathology, University Medical Centre Mannheim, University of Heidelberg, 68305 Mannheim, Germany; (C.S.); (A.M.); (D.H.)
| | - Ulrich Ronellenfitsch
- Department of Abdominal, Vascular, and Endocrine Surgery, University Hospital Halle (Saale), 06120 Halle (Saale), Germany;
| |
Collapse
|
8
|
Vyse S, Thway K, Huang PH, Jones RL. Next-generation sequencing for the management of sarcomas with no known driver mutations. Curr Opin Oncol 2021; 33:315-322. [PMID: 33927108 DOI: 10.1097/cco.0000000000000741] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
PURPOSE OF REVIEW Next-generation sequencing (NGS) has enabled fast, high-throughput nucleotide sequencing and has begun to be implemented into clinical practice for genomic-guided precision medicine in various cancer types. This review will discuss recent evidence that highlights opportunities for NGS to improve outcomes in sarcomas that have complex genomic profiles with no known driver mutations. RECENT FINDINGS Global genomic signatures detectable by NGS including tumour mutational burden and microsatellite instability have potential as biomarkers for response to immunotherapy in certain sarcoma subtypes including angiosarcomas. Identification of hallmarks associated with 'BRCAness' and homologous recombination repair defects in leiomyosarcomas and osteosarcomas may predict sensitivity to poly(adenosine diphosphate-ribose) polymerase (PARP) inhibitors. Lastly, the use of NGS for evaluating cancer predisposition in sarcomas may be useful for early detection, screening and surveillance. SUMMARY Currently, the implementation of NGS for every sarcoma patient is not practical or useful. However, adopting NGS as a complementary approach in sarcomas with complex genomics and those with limited treatment options has the potential to deliver precision medicine to a subgroup of patients, with novel therapies such as immune checkpoint and PARP inhibitors. Moving forward, molecular tumour boards incorporating multidisciplinary teams of pathologists, oncologists and genomic specialists to interpret NGS data will complement existing tools in diagnosis and treatment decision making in sarcoma patients.
Collapse
Affiliation(s)
- Simon Vyse
- Division of Molecular Pathology, The Institute of Cancer Research
| | - Khin Thway
- Sarcoma Unit, The Royal Marsden NHS Foundation Trust and Institute of Cancer Research, London, UK
| | - Paul H Huang
- Division of Molecular Pathology, The Institute of Cancer Research
| | - Robin L Jones
- Sarcoma Unit, The Royal Marsden NHS Foundation Trust and Institute of Cancer Research, London, UK
| |
Collapse
|
9
|
Pillozzi S, Bernini A, Palchetti I, Crociani O, Antonuzzo L, Campanacci D, Scoccianti G. Soft Tissue Sarcoma: An Insight on Biomarkers at Molecular, Metabolic and Cellular Level. Cancers (Basel) 2021; 13:cancers13123044. [PMID: 34207243 PMCID: PMC8233868 DOI: 10.3390/cancers13123044] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 06/13/2021] [Accepted: 06/14/2021] [Indexed: 12/18/2022] Open
Abstract
Simple Summary Soft tissue sarcoma is a rare mesenchymal malignancy. Despite the advancements in the fields of radiology, pathology and surgery, these tumors often recur locally and/or with metastatic disease. STS is considered to be a diagnostic challenge due to the large variety of histological subtypes with clinical and histopathological characteristics which are not always distinct. One of the important clinical problems is a lack of useful biomarkers. Therefore, the discovery of biomarkers that can be used to detect tumors or predict tumor response to chemotherapy or radiotherapy could help clinicians provide more effective clinical management. Abstract Soft tissue sarcomas (STSs) are a heterogeneous group of rare tumors. Although constituting only 1% of all human malignancies, STSs represent the second most common type of solid tumors in children and adolescents and comprise an important group of secondary malignancies. Over 100 histologic subtypes have been characterized to date (occurring predominantly in the trunk, extremity, and retroperitoneum), and many more are being discovered due to molecular profiling. STS mortality remains high, despite adjuvant chemotherapy. New prognostic stratification markers are needed to help identify patients at risk of recurrence and possibly apply more intensive or novel treatments. Recent scientific advancements have enabled a more precise molecular characterization of sarcoma subtypes and revealed novel therapeutic targets and prognostic/predictive biomarkers. This review aims at providing a comprehensive overview of the most relevant cellular, molecular and metabolic biomarkers for STS, and highlight advances in STS-related biomarker research.
Collapse
Affiliation(s)
- Serena Pillozzi
- Medical Oncology Unit, Careggi University Hospital, Largo Brambilla 3, 50134 Florence, Italy;
- Correspondence:
| | - Andrea Bernini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy;
| | - Ilaria Palchetti
- Department of Chemistry Ugo Schiff, University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Italy;
| | - Olivia Crociani
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy;
| | - Lorenzo Antonuzzo
- Medical Oncology Unit, Careggi University Hospital, Largo Brambilla 3, 50134 Florence, Italy;
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy;
| | - Domenico Campanacci
- Department of Health Science, University of Florence, Largo Brambilla 3, 50134 Florence, Italy;
| | - Guido Scoccianti
- Department of Orthopaedic Oncology and Reconstructive Surgery, University of Florence, Careggi University Hospital, Largo Brambilla 3, 50134 Florence, Italy;
| |
Collapse
|
10
|
Haas RL, Floot BGJ, Scholten AN, van der Graaf WTA, van Houdt W, Schrage Y, van de Ven M, Bovée JVMG, van Coevorden F, Vens C. Cellular Radiosensitivity of Soft Tissue Sarcoma. Radiat Res 2021; 196:23-30. [PMID: 33914890 DOI: 10.1667/rade-20-00226.1] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 03/19/2021] [Indexed: 11/03/2022]
Abstract
Currently, all soft tissue sarcomas (STS) are irradiated by the same regimen, disregarding possible subtype-specific radiosensitivities. To gain further insight, cellular radiosensitivity was investigated in a panel of sarcoma cell lines. Fourteen sarcoma cell lines, derived from synovial sarcoma, leiomyosarcoma, fibrosarcoma and liposarcoma origin, were submitted to clonogenic survival assays. Cells were irradiated with single doses from 1-8 Gy and surviving fraction (SF) was calculated from the resulting response data. Alpha/beta (α/β) ratios were inferred from radiation-response curves using the linear-quadratic (LQ)-model. Cellular radiosensitivities varied largely in this panel, indicating a considerable degree of heterogeneity. Surviving fraction after 2 Gy (SF2) ranged from 0.27 to 0.76 with evidence of a particular radiosensitive phenotype in only few cell lines. D37% on the mean data was 3.4 Gy and the median SF2 was 0.52. The median α/β was 4.9 Gy and in six cell lines the α/β was below 4 Gy. A fairly homogeneous radiation response was observed in myxoid liposarcoma cell lines with SF2 between 0.64 and 0.67. Further comparing sarcomas of different origin, synovial sarcomas, as a group, showed the lowest SF2 values (mean 0.35) and was significantly more radiosensitive than myxoid liposarcomas and leiomyosarcomas (P = 0.0084 and 0.024, respectively). This study demonstrates a broad spectrum of radiosensitivities across STS cell lines and reveals subtype-specific radiation responses. The particular cellular radiosensitivity of synovial sarcoma cells supports consideration of the different sarcoma entities in clinical studies that aim to optimize sarcoma radiotherapy.
Collapse
Affiliation(s)
- R L Haas
- Department of Radiotherapy, The Netherlands Cancer Insititute, Amsterdam, The Netherlands
| | - B G J Floot
- Department of Surgical Oncology, The Netherlands Cancer Insititute, Amsterdam, The Netherlands
| | - A N Scholten
- Department of Radiotherapy, The Netherlands Cancer Insititute, Amsterdam, The Netherlands
| | - W T A van der Graaf
- Division of Cell Biology, The Netherlands Cancer Insititute, Amsterdam, The Netherlands
| | - W van Houdt
- Department of Medical Oncology, The Netherlands Cancer Insititute, Amsterdam, The Netherlands
| | - Y Schrage
- Department of Medical Oncology, The Netherlands Cancer Insititute, Amsterdam, The Netherlands
| | - M van de Ven
- Preclinical Intervention Unit, Mouse Clinic for Cancer and Aging (MCCA), The Netherlands Cancer Insititute, Amsterdam, The Netherlands
| | - J V M G Bovée
- Department of Pathology, Leiden University Medical Centre, Leiden, the Netherlands
| | - F van Coevorden
- Department of Medical Oncology, The Netherlands Cancer Insititute, Amsterdam, The Netherlands
| | - C Vens
- Department of Radiotherapy, The Netherlands Cancer Insititute, Amsterdam, The Netherlands.,Department of Surgical Oncology, The Netherlands Cancer Insititute, Amsterdam, The Netherlands
| |
Collapse
|
11
|
Jin G, Wang C, Jia D, Qian W, Yin C, Wang D, Yang Q, Li T, Zheng A. Next Generation Sequencing Reveals Pathogenic and Actionable Genetic Alterations of Soft Tissue Sarcoma in Chinese Patients: A Single Center Experience. Technol Cancer Res Treat 2021; 20:15330338211068964. [PMID: 34939467 PMCID: PMC8721396 DOI: 10.1177/15330338211068964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 11/11/2021] [Accepted: 12/07/2021] [Indexed: 11/16/2022] Open
Abstract
Background: Next generation sequencing (NGS) has systematically investigated the genomic landscape of soft tissue sarcoma (STS) in Western patients, but few reports have described the utility of NGS in identifying pathogenic and targetable mutations in Asian patients. Methods: We review our single center experience of identifying the genomic profile and feasible genetic mutations in 65 Chinese patients with STS by NGS. Results: On average, 3.35 mutations were identified per patient (range, 0-28), and at least one mutation could be detected in 95.4% (62/65) of patients. TP53, MDM2, CDK4, KDR, and NF1 were the most frequent mutation genes in Chinese STS patients. Actionable mutations were discovered in 36.9% (24/65) of patients, and clinical benefit was achieved in 4 patients treated with corresponding molecular targeted therapies. Conclusions: Our study describes the mutation profile of Chinese STS patients by a single center experience. Some patients have achieved improved clinical outcomes by adopting treatment based on the results of genetic testing. NGS may affect clinical decision-making as a routine clinical test for patients with STS.
Collapse
Affiliation(s)
- Gu Jin
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | | | - Dongdong Jia
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Wenkang Qian
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | | | | | | | - Tao Li
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Aiwen Zheng
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| |
Collapse
|
12
|
Mas A, Simón C. Molecular differential diagnosis of uterine leiomyomas and leiomyosarcomas. Biol Reprod 2020; 101:1115-1123. [PMID: 30184111 DOI: 10.1093/biolre/ioy195] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 08/13/2018] [Accepted: 08/30/2018] [Indexed: 12/18/2022] Open
Abstract
Uterine leiomyomas (LM) and leiomyosarcomas (LMS) are considered biologically unrelated tumors due to their cytogenetic and molecular disparity. Yet, these tumors share morphological and molecular characteristics that cannot be differentiated through current clinical diagnostic tests, and thus cannot be definitively classified as benign or malignant until surgery. Newer approaches are needed for the identification of these tumors, as has been done for other tissues. The application of next generation sequencing enables the detection of new mutations that, when coupled to machine learning bioinformatic tools, advances our understanding of chromosomal instability. These approaches in the context of LM and LMS could allow the discovery of genetic variants and possible genomic markers. Additionally, the potential clinical utility of circulating cell-free tumor DNA could revolutionize the noninvasive detection and monitoring of these tumors. Here, we seek to provide a perspective on the molecular background of LM and LMS, recognizing their distinct molecular features that may lead to improved diagnosis and personalized treatments, which would have a measurable impact on women's reproductive health.
Collapse
Affiliation(s)
- Aymara Mas
- Reproductive Medicine Research Group, La Fe Health Research Institute, Valencia, Spain.,Igenomix Foundation/Instituto de Investigación Sanitaria Hospital Clínico (INCLIVA), Valencia, Spain
| | - Carlos Simón
- Igenomix Foundation/Instituto de Investigación Sanitaria Hospital Clínico (INCLIVA), Valencia, Spain.,Department of Pediatrics, Obstetrics and Gynecology, University of Valencia, Valencia, Spain
| |
Collapse
|
13
|
Gómez J, Tsagozis P. Multidisciplinary treatment of soft tissue sarcomas: An update. World J Clin Oncol 2020; 11:180-189. [PMID: 32355640 PMCID: PMC7186235 DOI: 10.5306/wjco.v11.i4.180] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 03/13/2020] [Accepted: 03/22/2020] [Indexed: 02/06/2023] Open
Abstract
Standard treatment for soft tissue sarcoma, based on complete surgical resection with or without adjuvant radiotherapy and chemotherapy, has not substantially changed during the last several decades. Nevertheless, recent advances have contributed to considerable improvement in the management of these patients; for example, new magnetic resonance imaging sequences such as diffusion-weighted imaging and magnetic resonance imaging radiomics can better assess tumor extension and even estimate its grade. Detection of circulating genetic material (liquid biopsy) and next-generation sequencing are powerful techniques for genetic analysis, which will increase our understanding of the underlying molecular mechanisms and may reveal potential therapeutic targets. The role of chemotherapy in non-metastatic disease is still controversial, and there is a need to identify patients who really benefit from this treatment. Novel chemotherapeutic regimens have entered clinical praxis and can change the outcome of patients with metastatic disease. Advances in radiotherapy have helped decrease local adverse effects and sustain good local control of the disease. The following report provides an updated view of the diagnosis, treatment, and future perspectives on the management of patients with soft tissue sarcomas.
Collapse
Affiliation(s)
- Jorge Gómez
- Department of Orthopedic Surgery, Clínica Universidad de Navarra, University of Navarra, Pamplona 31008, Spain
| | - Panagiotis Tsagozis
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm 17176, Sweden
- Muskuloskeletal Tumour Service, Karolinska University Hospital, Stockholm 17176, Sweden
| |
Collapse
|
14
|
Penel N, Lebellec L, Blay JY, Robin YM. Overview of « druggable » alterations by histological subtypes of sarcomas and connective tissue intermediate malignancies. Crit Rev Oncol Hematol 2020; 150:102960. [PMID: 32320927 DOI: 10.1016/j.critrevonc.2020.102960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 10/24/2022] Open
Abstract
We summarize herein the literature data about molecular targeted therapies in sarcomas and conjunctive tissue intermediate malignancies. For each clinical setting, the level of evidence, the mechanism of action and the target are described. The two major axes include (i) identification of subgroups of tumors with druggable alteration irrespective of the histological diagnosis (e.g. NTRK), and (ii) druggable target of pathway related to the physiopathology of the tumor: denosumab and bone giant cell tumor, imatinib and soft tissue giant cell tumor, mTOR inhibitor and PECOMA.
Collapse
Affiliation(s)
- Nicolas Penel
- Department of Medical Oncology, Centre Oscar Lambret, Lille, France; Lille University, Medical School, Lille, France.
| | - Loïc Lebellec
- Lille University, Medical School, Lille, France; Medical Oncology Unit, Dron Hospital, Tourcoing, France
| | - Jean-Yves Blay
- Department of Medicine, Centre Leon Bérard, Lyon, France; Claude Bernard University, Medical School Lyon, France
| | - Yves-Marie Robin
- Biopathology department, Centre Oscar Lambret, Lille, France; Lille University, Inserm U1192, Laboratoire « Protéomique, Réponse Inflammatoire et Spectrométrie de Masse » (PRISM), Villeneuve d'Ascq, France
| |
Collapse
|
15
|
Abstract
Pediatric musculoskeletal tumors can arise in both bone and soft tissues. The overwhelming majority of these are benign; however, rarely, malignant neoplasms do occur. These are collectively termed sarcomas, indicating their mesenchymal origin. Sarcoma management requires careful adherence to the well-described tenets of tumor management. This article summarizes the basic principles and recent advances in the management of soft tissue and bone tumors.
Collapse
Affiliation(s)
- Amit Singla
- Department of Orthopaedic Surgery, Montefiore Medical Center, 3400 Bainbridge Avenue, 6th Floor, Bronx, NY 10467, USA.
| | - David S Geller
- Department of Orthopaedic Surgery, Montefiore Medical Center, 3400 Bainbridge Avenue, 6th Floor, Bronx, NY 10467, USA
| |
Collapse
|
16
|
Mas A, Alonso R, Garrido-Gómez T, Escorcia P, Montero B, Jiménez-Almazán J, Martín J, Pellicer N, Monleón J, Simón C. The differential diagnoses of uterine leiomyomas and leiomyosarcomas using DNA and RNA sequencing. Am J Obstet Gynecol 2019; 221:320.e1-320.e23. [PMID: 31121144 DOI: 10.1016/j.ajog.2019.05.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/10/2019] [Accepted: 05/14/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Although uterine leiomyomas and leiomyosarcomas are considered biologically unrelated tumors, they share morphologic and histologic characteristics that complicate their differential diagnosis. The long-term therapeutic option for leiomyoma is laparoscopic myomectomy with morcellation, particularly for patients who wish to preserve their fertility. However, because of the potential dissemination of undiagnosed or hidden leiomyosarcoma from morcellation, there is a need to develop a preoperative assessment of malignancy risk. OBJECTIVE Through an integrated comparative genomic and transcriptomic analysis, we aim to identify differential genetic targets in leiomyomas vs leiomyosarcomas using next-generation sequencing as the first step toward preoperative differential diagnosis. STUDY DESIGN Targeted sequencing of DNA and RNA coding regions for solid tumor-associated genes was performed on formalin-fixed paraffin-embedded samples from 13 leiomyomas and 13 leiomyosarcoma cases. DNA sequencing was used to identify copy number variations, single-nucleotide variants, and small insertions/deletions. RNA sequencing was used to identify gene fusions, splice variants, and/or differential gene expression profiles. RESULTS In leiomyosarcomas, tumor mutation burden was higher in terms of copy number variations, single nucleotide variants, small insertions/deletions, and gene fusions compared with leiomyomas. For copy number variations, 20 genes were affected by deletions in leiomyosarcomas, compared with 6 observed losses in leiomyomas. Gains (duplications) were identified in 19 genes in leiomyosarcomas, but only 3 genes in leiomyomas. The most common mutations (single-nucleotide variants and insertions/deletions) for leiomyosarcomas were identified in 105 genes of all analyzed leiomyosarcomas; 82 genes were affected in leiomyomas. Of note, 1 tumor previously diagnosed as leiomyosarcoma was established as inflammatory myofibroblastic tumor along this study with a novel ALK-TNS1 fusion. Finally, a differential transcriptomic profile was observed for 11 of 55 genes analyzed in leiomyosarcomas; 8.5% of initially diagnosed leiomyosarcomas showed high-confidence, novel gene fusions that were associated with these tumors. CONCLUSION Through integrated comparative genomic and transcriptomic analyses, we identified novel differential genetic targets that potentially differentiate leiomyosarcomas and leiomyomas. This provides a new insight into the differential diagnosis of these myometrial tumors.
Collapse
|
17
|
Pei J, Zhao X, Patchefsky AS, Flieder DB, Talarchek JN, Testa JR, Wei S. Clinical application of RNA sequencing in sarcoma diagnosis: An institutional experience. Medicine (Baltimore) 2019; 98:e16031. [PMID: 31232935 PMCID: PMC6636967 DOI: 10.1097/md.0000000000016031] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Accurate diagnoses of sarcoma are sometimes challenging on conventional histomorphology and immunophenotype. Many specific genetic aberrations including chromosomal translocations have been identified in various sarcomas, which can be detected by fluorescence in situ hybridization and polymerase chain reaction analysis. Next-generation sequencing-based RNA sequencing can screen multiple sarcoma-specific chromosome translocations/fusion genes in 1 test, which is especially useful for sarcoma without obvious differentiation. In this report, we utilized RNA sequencing on formalin-fixed paraffin-embedded (FFPE) specimens to investigate the possibility of diagnosing sarcomas by identifying disease-specific fusion genes. Targeted RNA sequencing was performed on 6 sarcoma cases. The expected genetic alterations (clear cell sarcoma/EWSR1-ATF1, Ewing sarcoma/EWSR1-FLI1, myxoid liposarcoma/DDIT3-FUS) in four cases were detected and confirmed by secondary tests. Interestingly, three SS18 fusion genes (SS18-SSX2B, SS18-SSX2, and SS18-SSX4) were identified in a synovial sarcoma case. A rare fusion gene (EWSR1-PATZ1) was identified in a morphologically challenging case; which enabled us to establish the diagnosis of low grade glioneural tumor. In conclusion, RNA sequencing on FFPE specimen is a reliable method in establishing the diagnosis of sarcoma in daily practice.
Collapse
Affiliation(s)
| | - Xiaofeng Zhao
- Department of Pathology and Laboratory Medicine, Temple University Hospital
| | | | | | | | - Joseph R. Testa
- Genomics Facility, Fox Chase Cancer Center
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA
| | | |
Collapse
|
18
|
Yazawa Y. Recent impacts of cytogenetic studies in musculoskeletal oncology. J Orthop Sci 2019; 24:385-386. [PMID: 30879935 DOI: 10.1016/j.jos.2019.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 03/01/2019] [Indexed: 02/09/2023]
Affiliation(s)
- Yasuo Yazawa
- Orthopaedic Oncology and Surgery, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, Saitama, 350-1298, Japan.
| |
Collapse
|
19
|
Carmagnani Pestana R, Groisberg R, Roszik J, Subbiah V. Precision Oncology in Sarcomas: Divide and Conquer. JCO Precis Oncol 2019; 3:PO.18.00247. [PMID: 32914012 PMCID: PMC7446356 DOI: 10.1200/po.18.00247] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2019] [Indexed: 12/18/2022] Open
Abstract
Sarcomas are a heterogeneous group of rare malignancies that exhibit remarkable heterogeneity, with more than 50 subtypes recognized. Advances in next-generation sequencing technology have resulted in the discovery of genetic events in these mesenchymal tumors, which in addition to enhancing understanding of the biology, have opened up avenues for molecularly targeted therapy and immunotherapy. This review focuses on how incorporation of next-generation sequencing has affected drug development in sarcomas and strategies for optimizing precision oncology for these rare cancers. In a significant percentage of soft tissue sarcomas, which represent up to 40% of all sarcomas, specific driver molecular abnormalities have been identified. The challenge to evaluate these mutations across rare cancer subtypes requires the careful characterization of these genetic alterations to further define compelling drivers with therapeutic implications. Novel models of clinical trial design also are needed. This shift would entail sustained efforts by the sarcoma community to move from one-size-fits-all trials, in which all sarcomas are treated similarly, to divide-and-conquer subtype-specific strategies.
Collapse
Affiliation(s)
| | - Roman Groisberg
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jason Roszik
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Vivek Subbiah
- The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
20
|
Drug sensitivity testing on patient-derived sarcoma cells predicts patient response to treatment and identifies c-Sarc inhibitors as active drugs for translocation sarcomas. Br J Cancer 2019; 120:435-443. [PMID: 30745580 PMCID: PMC6462037 DOI: 10.1038/s41416-018-0359-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 10/26/2018] [Accepted: 11/22/2018] [Indexed: 11/17/2022] Open
Abstract
Background Heterogeneity and low incidence comprise the biggest challenge in sarcoma diagnosis and treatment. Chemotherapy, although efficient for some sarcoma subtypes, generally results in poor clinical responses and is mostly recommended for advanced disease. Specific genomic aberrations have been identified in some sarcoma subtypes but few of them can be targeted with approved drugs. Methods We cultured and characterised patient-derived sarcoma cells and evaluated their sensitivity to 525 anti-cancer agents including both approved and non-approved drugs. In total, 14 sarcomas and 5 healthy mesenchymal primary cell cultures were studied. The sarcoma biopsies and derived cells were characterised by gene panel sequencing, cancer driver gene expression and by detecting specific fusion oncoproteins in situ in sarcomas with translocations. Results Soft tissue sarcoma cultures were established from patient biopsies with a success rate of 58%. The genomic profile and drug sensitivity testing on these samples helped to identify targeted inhibitors active on sarcomas. The cSrc inhibitor Dasatinib was identified as an active drug in sarcomas carrying chromosomal translocations. The drug sensitivity of the patient sarcoma cells ex vivo correlated with the response to the former treatment of the patient. Conclusions Our results show that patient-derived sarcoma cells cultured in vitro are relevant and practical models for genotypic and phenotypic screens aiming to identify efficient drugs to treat sarcoma patients with poor treatment options.
Collapse
|
21
|
Groisberg R, Hong DS, Holla V, Janku F, Piha-Paul S, Ravi V, Benjamin R, Kumar Patel S, Somaiah N, Conley A, Ali SM, Schrock AB, Ross JS, Stephens PJ, Miller VA, Sen S, Herzog C, Meric-Bernstam F, Subbiah V. Clinical genomic profiling to identify actionable alterations for investigational therapies in patients with diverse sarcomas. Oncotarget 2018; 8:39254-39267. [PMID: 28424409 PMCID: PMC5503611 DOI: 10.18632/oncotarget.16845] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Accepted: 03/08/2017] [Indexed: 12/22/2022] Open
Abstract
Background There are currently no United States Food and Drug Administration approved molecularly matched therapies for sarcomas except gastrointestinal stromal tumors. Complicating this is the extreme diversity, heterogeneity, and rarity of these neoplasms. Few therapeutic options exist for relapsed and refractory sarcomas. In clinical practice many oncologists refer patients for genomic profiling hoping for guidance on treatment options after standard therapy. However, a systematic analysis of actionable mutations has yet to be completed. We analyzed genomic profiling results in patients referred to MD Anderson Cancer Center with advanced sarcomas to elucidate the frequency of potentially actionable genomic alterations in this population. Methods We reviewed charts of patients with advanced sarcoma who were referred to investigational cancer therapeutics department and had CLIA certified comprehensive genomic profiling (CGP) of 236 or 315 cancer genes in at least 50ng of DNA. Actionable alterations were defined as those identifying anti-cancer drugs on the market, in registered clinical trials, or in the Drug-Gene Interaction Database. Results Among the 102 patients analyzed median age was 45.5 years (range 8-76), M: F ratio 48:54. The most common subtypes seen in our study were leiomyosarcoma (18.6%), dedifferentiated liposarcoma (11%), osteosarcoma (11%), well-differentiated liposarcoma (7%), carcinosarcoma (6%), and rhabdomyosarcoma (6%). Ninety-five out of 102 patients (93%) had at least one genomic alteration identified with a mean of six mutations per patient. Of the 95 biopsy samples with identifiable genomic alterations, the most commonly affected genes were TP53 (31.4%), CDK4 (23.5%), MDM2 (21.6%), RB1 (18.6%), and CDKN2A/B (13.7%). Notable co-segregating amplifications included MDM2-CDK4 and FRS2-FGF. Sixteen percent of patients received targeted therapy based on CGP of which 50% had at least stable disease. Conclusions Incorporating CGP into sarcoma management may allow for more precise diagnosis and sub-classification of this diverse and rare disease, as well as personalized matching of patients to targeted therapies such as those available in basket clinical trials.
Collapse
Affiliation(s)
- Roman Groisberg
- Department of Investigational Cancer Therapeutics (A Phase I Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA.,Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - David S Hong
- Department of Investigational Cancer Therapeutics (A Phase I Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Vijaykumar Holla
- Khalifa Institute for Personalized Cancer Therapy (IPCT), The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Filip Janku
- Department of Investigational Cancer Therapeutics (A Phase I Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Sarina Piha-Paul
- Department of Investigational Cancer Therapeutics (A Phase I Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Vinod Ravi
- Department of Sarcoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Robert Benjamin
- Department of Sarcoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Shreyas Kumar Patel
- Department of Sarcoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Neeta Somaiah
- Department of Sarcoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Anthony Conley
- Department of Sarcoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Siraj M Ali
- Foundation Medicine Inc, Cambridge, Massachusetts 02139, USA
| | - Alexa B Schrock
- Foundation Medicine Inc, Cambridge, Massachusetts 02139, USA
| | - Jeffrey S Ross
- Foundation Medicine Inc, Cambridge, Massachusetts 02139, USA
| | | | | | - Shiraj Sen
- Department of Investigational Cancer Therapeutics (A Phase I Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA.,Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Cynthia Herzog
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics (A Phase I Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Vivek Subbiah
- Department of Investigational Cancer Therapeutics (A Phase I Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
22
|
Ai JW, Li Y, Cheng Q, Cui P, Wu HL, Xu B, Zhang WH. Diagnosis of local hepatic tuberculosis through next-generation sequencing: Smarter, faster and better. Clin Res Hepatol Gastroenterol 2018; 42:178-181. [PMID: 29759945 DOI: 10.1016/j.clinre.2018.04.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 04/19/2018] [Indexed: 02/04/2023]
Abstract
BACKGROUND A 45-year-old man who complained of continuous fever and multiple hepatic masses was admitted to our hospital. Repeated MRI manifestations were similar while each radiological report suggested contradictory diagnosis pointing to infections or malignances respectively. Pathologic examination of the liver tissue showed no direct evidence of either infections or tumor. We performed next-generation sequencing on the liver tissue and peripheral blood to further investigate the possible etiology. METHODS High throughput sequencing was performed on the liver lesion tissues using BGISEQ-100 platform, and data was mapped to the Microbial Genome Databases after filtering low quality data and human reads. RESULTS We identified a total of 299 sequencing reads of Mycobacterium tuberculosis (M. tuberculosis) complex sequences from the liver tissue, including 8, 229 of 4,424,435 of the M. tuberculosis nucleotide sequences, and Mycobacterium africanum, Mycobacterium bovis, and Mycobacterium canettii were also detected due to the 99.9% identical rate among these strains. No specific Mycobacterial tuberculosis nucleotide sequence was detected in the sample of peripheral blood. Patient's symptom quickly recovered after anti-tuberculosis treatment and repeated Ziehl-Neelsen staining of the liver tissue finally identified small numbers of positive bacillus. CONCLUSIONS The diagnosis of this patient was difficult to establish before the next-generation sequencing because of contradictive radiological results and negative pathological findings. More sensitive diagnostic methods are urgently needed. This is the first case reporting hepatic tuberculosis confirmed by the next-generation sequencing, and marks the promising potential of the application of the next-generation sequencing in the diagnosis of hepatic lesions with unknown etiology.
Collapse
Affiliation(s)
- Jing-Wen Ai
- Department of infectious disease, Huashan Hospital of Fudan University, Shanghai, China.
| | - Yang Li
- Department of infectious disease, Huashan Hospital of Fudan University, Shanghai, China.
| | - Qi Cheng
- Department of infectious disease, Huashan Hospital of Fudan University, Shanghai, China.
| | - Peng Cui
- Department of infectious disease, Huashan Hospital of Fudan University, Shanghai, China.
| | - Hong-Long Wu
- Binhai Genomics Institute, Tianjin Translational Genomics Center, BGI-Tianjin, BGI-Shenzhen, Tianjin, China; BGI-Shenzhen, China.
| | - Bin Xu
- Department of infectious disease, Huashan Hospital of Fudan University, Shanghai, China.
| | - Wen-Hong Zhang
- Department of infectious disease, Huashan Hospital of Fudan University, Shanghai, China.
| |
Collapse
|
23
|
Chan JYK, Poon PHY, Zhang Y, Ng CWK, Piao WY, Ma M, Yip KY, Chan ABW, Lui VWY. Case Report: Exome sequencing reveals recurrent RETSAT mutations and a loss-of-function POLDIP2 mutation in a rare undifferentiated tongue sarcoma. F1000Res 2018; 7:499. [PMID: 29862022 PMCID: PMC5954348 DOI: 10.12688/f1000research.14383.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/20/2018] [Indexed: 01/08/2023] Open
Abstract
Soft tissue sarcoma of the tongue represents a very rare head and neck cancer with connective tissue features, and the genetics underlying this rare cancer are largely unknown. There are less than 20 cases reported in the literature thus far. Here, we reported the first whole-exome characterization (>×200 depth) of an undifferentiated sarcoma of the tongue in a 31-year-old male. Even with a very good sequencing depth, only 19 nonsynonymous mutations were found, indicating a relatively low mutation rate of this rare cancer (lower than that of human papillomavirus (HPV)-positive head and neck cancer). Yet, among the few genes that are somatically mutated in this HPV-negative undifferentiated tongue sarcoma, a noticeable deleterious frameshift mutation (with a very high allele frequency of >93%) of a gene for DNA replication and repair, namely
POLDIP2 (DNA polymerase delta interacting protein 2), and two recurrent mutations of the adipogenesis and adipocyte differentiation gene
RETSAT (retinol saturase), were identified. Thus, somatic events likely affecting adipogenesis and differentiation, as well as potential stem mutations to
POLDIP2, may be implicated in the formation of this rare cancer. This identified somatic whole-exome sequencing profile appears to be distinct from that of other reported adult sarcomas from The Cancer Genome Atlas, suggesting a potential unique genetic profile for this rare sarcoma of the tongue. Interestingly, this low somatic mutation rate is unexpectedly found to be accompanied by multiple tumor protein p53 and
NOTCH1 germline mutations of the patient’s blood DNA. This may explain the very early age of onset of head and neck cancer, with likely hereditary predisposition. Our findings are, to our knowledge, the first to reveal a unique genetic profile of this very rare undifferentiated sarcoma of the tongue.
Collapse
Affiliation(s)
- Jason Y K Chan
- Department of Otorhinolaryngology, Head and Neck Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Peony Hiu Yan Poon
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Yong Zhang
- Department of Computer Science and Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Cherrie W K Ng
- Department of Otorhinolaryngology, Head and Neck Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Wen Ying Piao
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Meng Ma
- Chongqing Medical University, Chongqing, China
| | - Kevin Y Yip
- Department of Computer Science and Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Amy B W Chan
- Department of Anatomical and Cellular Pathology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Vivian Wai Yan Lui
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
24
|
Sarcoma Spheroids and Organoids-Promising Tools in the Era of Personalized Medicine. Int J Mol Sci 2018; 19:ijms19020615. [PMID: 29466296 PMCID: PMC5855837 DOI: 10.3390/ijms19020615] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 02/13/2018] [Accepted: 02/16/2018] [Indexed: 02/06/2023] Open
Abstract
Cancer treatment is rapidly evolving toward personalized medicine, which takes into account the individual molecular and genetic variability of tumors. Sophisticated new in vitro disease models, such as three-dimensional cell cultures, may provide a tool for genetic, epigenetic, biomedical, and pharmacological research, and help determine the most promising individual treatment. Sarcomas, malignant neoplasms originating from mesenchymal cells, may have a multitude of genomic aberrations that give rise to more than 70 different histopathological subtypes. Their low incidence and high level of histopathological heterogeneity have greatly limited progress in their treatment, and trials of clinical sarcoma are less frequent than trials of other carcinomas. The main advantage of 3D cultures from tumor cells or biopsy is that they provide patient-specific models of solid tumors, and they overcome some limitations of traditional 2D monolayer cultures by reflecting cell heterogeneity, native histologic architectures, and cell-extracellular matrix interactions. Recent advances promise that these models can help bridge the gap between preclinical and clinical research by providing a relevant in vitro model of human cancer useful for drug testing and studying metastatic and dormancy mechanisms. However, additional improvements of 3D models are expected in the future, specifically the inclusion of tumor vasculature and the immune system, to enhance their full ability to capture the biological features of native tumors in high-throughput screening. Here, we summarize recent advances and future perspectives of spheroid and organoid in vitro models of rare sarcomas that can be used to investigate individual molecular biology and predict clinical responses. We also highlight how spheroid and organoid culture models could facilitate the personalization of sarcoma treatment, provide specific clinical scenarios, and discuss the relative strengths and limitations of these models.
Collapse
|
25
|
Gao P, Seebacher NA, Hornicek F, Guo Z, Duan Z. Advances in sarcoma gene mutations and therapeutic targets. Cancer Treat Rev 2017; 62:98-109. [PMID: 29190505 DOI: 10.1016/j.ctrv.2017.11.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 10/31/2017] [Accepted: 11/01/2017] [Indexed: 12/14/2022]
Abstract
Sarcomas are rare and complex malignancies that have been associated with a poor prognostic outcome. Over the last few decades, traditional treatment with surgery and/or chemotherapy has not significantly improved outcomes for most types of sarcomas. In recent years, there have been significant advances in the understanding of specific gene mutations that are important in driving the pathogenesis and progression of sarcomas. Identification of these new gene mutations, using next-generation sequencing and advanced molecular techniques, has revealed a range of potential therapeutic targets. This, in turn, may lead to the development of novel agents targeted to different sarcoma subtypes. In this review, we highlight the advances made in identifying sarcoma gene mutations, including those of p53, RB, PI3K and IDH genes, as well as novel therapeutic strategies aimed at utilizing these mutant genes. In addition, we discuss a number of preclinical studies and ongoing early clinical trials in sarcoma targeting therapies, as well as gene editing technology, which may provide a better choice for sarcoma patient management.
Collapse
Affiliation(s)
- Peng Gao
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China; UCLA Orthopaedic Surgery, Sarcoma Biology Laboratory, 615 Charles E Young Dr. South, Biomedical Sciences Research Building, Room 410, Los Angeles, CA 90095, USA
| | - Nicole A Seebacher
- UCLA Orthopaedic Surgery, Sarcoma Biology Laboratory, 615 Charles E Young Dr. South, Biomedical Sciences Research Building, Room 410, Los Angeles, CA 90095, USA
| | - Francis Hornicek
- UCLA Orthopaedic Surgery, Sarcoma Biology Laboratory, 615 Charles E Young Dr. South, Biomedical Sciences Research Building, Room 410, Los Angeles, CA 90095, USA
| | - Zheng Guo
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| | - Zhenfeng Duan
- UCLA Orthopaedic Surgery, Sarcoma Biology Laboratory, 615 Charles E Young Dr. South, Biomedical Sciences Research Building, Room 410, Los Angeles, CA 90095, USA.
| |
Collapse
|
26
|
The Role of Next-Generation Sequencing in Sarcomas: Evolution From Light Microscope to Molecular Microscope. Curr Oncol Rep 2017; 19:78. [DOI: 10.1007/s11912-017-0641-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
27
|
Lewin J, Garg S, Lau BY, Dickson BC, Traub F, Gokgoz N, Griffin AM, Ferguson PC, Andrulis IL, Sim HW, Kamel-Reid S, Stockley TL, Siu LL, Wunder JS, Razak ARA. Identifying actionable variants using next generation sequencing in patients with a historical diagnosis of undifferentiated pleomorphic sarcoma. Int J Cancer 2017; 142:57-65. [PMID: 28891048 DOI: 10.1002/ijc.31039] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 07/19/2017] [Accepted: 08/24/2017] [Indexed: 12/22/2022]
Abstract
There are limited data regarding the molecular characterization of undifferentiated pleomorphic sarcomas (UPS; formerly malignant fibrous histiocytoma). This study aimed to investigate the utility of next generation sequencing (NGS) in UPS to identify subsets of patients who harbour actionable mutations. Patients diagnosed with UPS underwent pathological re-evaluation by a pathologist specializing in sarcoma. Tumor DNA was isolated from archived fresh frozen tissue samples and genotyped using NGS with the Illumina MiSeq TruSeq Amplicon Cancer Panel (48 genes, 212 amplicons). In total, 95 patients initially classified with UPS were identified. Following pathology re-review the histological subtypes were reclassified to include: Myxofibrosarcoma (MFS, N = 44); UPS(N = 18); and Others (N = 27; including undifferentiated spindle cell sarcoma (N = 15) and dedifferentiated liposarcoma (N = 6)). Seven cases were excluded from further analysis for other reasons. Baseline demographics of the finalized cohort (N = 88) showed a median age of 66 years (32-95), primarily with stage I-III disease (92%) and high-grade (86%) lesions. Somatic mutations were identified in 31 cases (35%)(Total mutations = 36: solitary mutation(n = 27); two mutations( =n = 3); three mutations(n = 1)). The most commonly identified mutations were in TP53 (n = 24), ATM (n = 3) and PIK3CA (n = 2). Three of 43 patients with MFS and one of 18 patients with UPS had clinically relevant mutations, mainly related to biomarkers of prediction of response; however few had targetable driver mutations. Somatic mutation status did not influence disease free or overall survival. Based on the small number of clinically relevant mutations, these data do not support the routine use of targeted NGS panels outside of research protocols in UPS.
Collapse
Affiliation(s)
- Jeremy Lewin
- Sarcoma Program, Mount Sinai Hospital, Toronto, Canada.,Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Canada
| | - Swati Garg
- Advanced Molecular Diagnostics Laboratory, Princess Margaret Cancer Centre, Toronto, Canada
| | - Beatrice Y Lau
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Canada
| | - Brendan C Dickson
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Canada
| | - Frank Traub
- Department of Orthopedic surgery, University Hospital Tuebingen, Erberhad Karls University Tuebingen, Germany
| | - Nalan Gokgoz
- Lunenfeld-Tanenbaum Research Institute, Toronto, Canada
| | | | - Peter C Ferguson
- Department of Surgery, Mount Sinai Hospital, University of Toronto, University Musculoskeletal Oncology Unit and Division of Orthopaedic Surgery, Toronto, Canada
| | - Irene L Andrulis
- Lunenfeld-Tanenbaum Research Institute, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Hao-Wen Sim
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Canada
| | - Suzanne Kamel-Reid
- Advanced Molecular Diagnostics Laboratory, Princess Margaret Cancer Centre, Toronto, Canada.,Department of Clinical Laboratory Genetics, Laboratory Medicine Program, University Health Network, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Candada.,Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Tracy L Stockley
- Advanced Molecular Diagnostics Laboratory, Princess Margaret Cancer Centre, Toronto, Canada.,Department of Clinical Laboratory Genetics, Laboratory Medicine Program, University Health Network, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Candada
| | - Lillian L Siu
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Canada
| | - Jay S Wunder
- Department of Surgery, Mount Sinai Hospital, University of Toronto, University Musculoskeletal Oncology Unit and Division of Orthopaedic Surgery, Toronto, Canada
| | - Albiruni R A Razak
- Sarcoma Program, Mount Sinai Hospital, Toronto, Canada.,Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Canada
| |
Collapse
|
28
|
Gaebler M, Silvestri A, Haybaeck J, Reichardt P, Lowery CD, Stancato LF, Zybarth G, Regenbrecht CRA. Three-Dimensional Patient-Derived In Vitro Sarcoma Models: Promising Tools for Improving Clinical Tumor Management. Front Oncol 2017; 7:203. [PMID: 28955656 PMCID: PMC5601986 DOI: 10.3389/fonc.2017.00203] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 08/21/2017] [Indexed: 12/12/2022] Open
Abstract
Over the past decade, the development of new targeted therapeutics directed against specific molecular pathways involved in tumor cell proliferation and survival has allowed an essential improvement in carcinoma treatment. Unfortunately, the scenario is different for sarcomas, a group of malignant neoplasms originating from mesenchymal cells, for which the main therapeutic approach still consists in the combination of surgery, chemotherapy, and radiation therapy. The lack of innovative approaches in sarcoma treatment stems from the high degree of heterogeneity of this tumor type, with more that 70 different histopathological subtypes, and the limited knowledge of the molecular drivers of tumor development and progression. Currently, molecular therapies are available mainly for the treatment of gastrointestinal stromal tumor, a soft-tissue malignancy characterized by an activating mutation of the tyrosine kinase KIT. Since the first application of this approach, a strong effort has been made to understand sarcoma molecular alterations that can be potential targets for therapy. The low incidence combined with the high level of histopathological heterogeneity makes the development of clinical trials for sarcomas very challenging. For this reason, preclinical studies are needed to better understand tumor biology with the aim to develop new targeted therapeutics. Currently, these studies are mainly based on in vitro testing, since cell lines, and in particular patient-derived models, represent a reliable and easy to handle tool for investigation. In the present review, we summarize the most important models currently available in the field, focusing in particular on the three-dimensional spheroid/organoid model. This innovative approach for studying tumor biology better represents tissue architecture and cell–cell as well as cell–microenvironment crosstalk, which are fundamental steps for tumor cell proliferation and survival.
Collapse
Affiliation(s)
- Manuela Gaebler
- HELIOS Klinikum Berlin-Buch GmbH, Department of Interdisciplinary Oncology, Berlin, Germany
| | | | - Johannes Haybaeck
- Medical Faculty, Department of Pathology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.,Institute of Pathology, Medical University Graz, Graz, Austria
| | - Peter Reichardt
- HELIOS Klinikum Berlin-Buch GmbH, Department of Interdisciplinary Oncology, Berlin, Germany
| | - Caitlin D Lowery
- Eli Lilly and Company, Oncology Translational Research, Lilly Corporate Center, Indianapolis, IN, United States
| | - Louis F Stancato
- Eli Lilly and Company, Oncology Translational Research, Lilly Corporate Center, Indianapolis, IN, United States
| | - Gabriele Zybarth
- cpo - Cellular Phenomics & Oncology Berlin-Buch GmbH, Berlin, Germany
| | | |
Collapse
|
29
|
Sellevold S, Peng Q, Fremstedal ASV, Berg K. Photochemical internalization (PCI) of bleomycin is equally effective in two dissimilar leiomyosarcoma xenografts in athymic mice. Photodiagnosis Photodyn Ther 2017; 20:95-106. [PMID: 28865875 DOI: 10.1016/j.pdpdt.2017.08.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 08/21/2017] [Accepted: 08/27/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND Photochemical internalization (PCI) is a novel technique for delivery of active macromolecules into cancerous cells, via light activation of a specific photosensitizer and a low dose systemic drug. Numerous pre-clinical studies and one clinical trial have confirmed the treatment potential in carcinomas. Soft tissue sarcomas are rare and generally resistant to radio- and chemotherapy. Due to treatment resistance and surgical morbidity in sarcoma care, we seek to increase knowledge on PCI effects in sarcomas by studying two different, but closely related leiomyosarcomas. METHODS MES-SA and SK-LMS-1 tumours were established in the leg muscles of athymic mice. Treatment effects after AlPcS2a-PCI of bleomycin, PCI with no drug (photodynamic therapy, PDT) and control groups were evaluated by: 1) assessment of tumour growth, 2) uptake of contrast agent during MRI and 3) histopathology. RESULTS PCI of bleomycin induced a similar and significant increase in time to reach the end point in both tumour models, while neither responded to AlPcS2a-PDT. In the MES-SA tumours PCI reduced the growth rate, while in the SK-LMS-1 tumours the growth was blocked for 12days followed by exponential growth close to that of untreated tumours. SK-LMS-1 tumours were more homogenously and better vascularized than MES-SA. After PCI the vascular shutdown was more complete in the SK-LMS-1 tumours than in the MES-SA tumours. CONCLUSIONS AlPcS2a-based PCI, but not PDT, induced significant tumour growth delay in the evaluated sarcomas. Cellular responsiveness to bleomycin and tumour vascularity are identified as predictive markers for PCI treatment effects.
Collapse
Affiliation(s)
- Simen Sellevold
- Division of Orthopaedic Surgery, Oslo University Hospital, Norway; Department of Radiation Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
| | - Qian Peng
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, Norway
| | - Ane Sofie Viset Fremstedal
- Department of Radiation Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
| | - Kristian Berg
- Department of Radiation Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway.
| |
Collapse
|
30
|
Al-Zaid T, Wang WL, Somaiah N, Lazar AJ. Molecular profiling of sarcomas: new vistas for precision medicine. Virchows Arch 2017; 471:243-255. [PMID: 28664413 DOI: 10.1007/s00428-017-2174-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 06/06/2017] [Accepted: 06/08/2017] [Indexed: 12/27/2022]
Abstract
Sarcoma is a large and heterogeneous group of malignant mesenchymal neoplasms with significant histological overlap. Accurate diagnosis can be challenging yet important for selecting the appropriate treatment approach and prognosis. The currently torrid pace of new genomic discoveries aids our classification and diagnosis of sarcomas, understanding of pathogenesis, development of new medications, and identification of alterations that predict prognosis and response to therapy. Unfortunately, demonstrating effective targets for precision oncology has been elusive in most sarcoma types. The list of potential targets greatly outnumbers the list of available inhibitors at the present time. This review will discuss the role of molecular profiling in sarcomas in general with emphasis on selected entities with particular clinical relevance.
Collapse
Affiliation(s)
- Tariq Al-Zaid
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Wei-Lien Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Boulevard-Unit 85, Houston, TX, 77030-4009, USA.,Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Sarcoma Research Center, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Neeta Somaiah
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alexander J Lazar
- Department of Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Boulevard-Unit 85, Houston, TX, 77030-4009, USA. .,Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. .,Sarcoma Research Center, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
31
|
Shim JH, Shin HT, Park J, Park JH, Lee JH, Yang JM, Kim DH, Jang KT, Lee DY. Mutational profiling of acral melanomas in Korean populations. Exp Dermatol 2017; 26:883-888. [DOI: 10.1111/exd.13321] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Joon Ho Shim
- Department of Dermatology; Samsung Medical Center; Sungkyunkwan University School of Medicine; Seoul Korea
- Samsung Genome Institute; Samsung Medical Center; Seoul Korea
| | - Hyun-Tae Shin
- Samsung Genome Institute; Samsung Medical Center; Seoul Korea
| | - Jiho Park
- Department of Health Sciences and Technology; Samsung Advanced Institute for Health Sciences and Technology; Sungkyunkwan University; Seoul Korea
| | - Ji-Hye Park
- Department of Dermatology; Samsung Medical Center; Sungkyunkwan University School of Medicine; Seoul Korea
| | - Jong-Hee Lee
- Department of Dermatology; Samsung Medical Center; Sungkyunkwan University School of Medicine; Seoul Korea
- Department of Medical Device Management & Research; Samsung Advanced Institute for Health Sciences and Technology; Sungkyunkwan University; Seoul Korea
| | - Jun-Mo Yang
- Department of Dermatology; Samsung Medical Center; Sungkyunkwan University School of Medicine; Seoul Korea
| | - Duk-Hwan Kim
- Department of Molecular Cell Biology; Samsung Biomedical Research Institute; Sungkyunkwan University School of Medicine; Suwon Korea
| | - Kee-Taek Jang
- Department of Pathology; Samsung Medical Center; Sungkyunkwan University School of Medicine; Seoul Korea
| | - Dong-Youn Lee
- Department of Dermatology; Samsung Medical Center; Sungkyunkwan University School of Medicine; Seoul Korea
| |
Collapse
|
32
|
Histology-Specific Uses of Tyrosine Kinase Inhibitors in Non-gastrointestinal Stromal Tumor Sarcomas. Curr Treat Options Oncol 2016; 17:11. [PMID: 26931561 DOI: 10.1007/s11864-015-0382-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
OPINION STATEMENT Adult sarcomas, especially those with metastatic or unresectable disease, have limited treatment options. Traditional chemotherapeutic options have been limited by poor response rates in patients with advanced sarcomas. The important clinical question is whether the success of targeted therapy in GIST can be extended to other sarcomas and also if preclinical data describing targets across this heterogeneous group of cancers can be translated to clinical efficacy of known and upcoming target specific agents. Multi-targeted tyrosine kinase inhibitors (TKI) such as pazopanib, sorafenib, sunutinib, cediranib have shown benefits across various histologies of soft tissue sarcoma as well as bone sarcomas. The efficacy of imatinib in Dermatofibrosarcoma Protruberans; sunitinib and cediranib in alveolar soft part sarcoma; and sorafenib and imatinib in chordomas have provided a treatment option of these rare tumors where no effective options existed. TKIs are being tested in combination with chemotherapy as well as radiation to improve response. Although traditional RECIST criteria may not adequately reflect response to these targeted agents, the studies have shown promise for the efficacy of TKIs across the spectrum of sarcomas. The integration of biomarker studies with clinical trials may help further identify responders beyond that defined by histology. With the current data, TKIs are being used both as first-line treatment and beyond in non-GIST sarcomas.
Collapse
|
33
|
Andersson C, Fagman H, Hansson M, Enlund F. Profiling of potential driver mutations in sarcomas by targeted next generation sequencing. Cancer Genet 2016; 209:154-60. [PMID: 26987750 DOI: 10.1016/j.cancergen.2016.02.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 01/19/2016] [Accepted: 02/08/2016] [Indexed: 11/19/2022]
Abstract
Comprehensive genetic profiling by massively parallel sequencing, commonly known as next generation sequencing (NGS), is becoming the foundation of personalized oncology. For sarcomas very few targeted treatments are currently in routine use. In clinical practice the preoperative diagnostic workup of soft tissue tumours largely relies on core needle biopsies. Although mostly sufficient for histopathological diagnosis, only very limited amounts of formalin fixated paraffin embedded tissue are often available for predictive mutation analysis. Targeted NGS may thus open up new possibilities for comprehensive characterization of scarce biopsies. We therefore set out to search for driver mutations by NGS in a cohort of 55 clinically and morphologically well characterized sarcomas using low input of DNA from formalin fixated paraffin embedded tissues. The aim was to investigate if there are any recurrent or targetable aberrations in cancer driver genes in addition to known chromosome translocations in different types of sarcomas. We employed a panel covering 207 mutation hotspots in 50 cancer-associated genes to analyse DNA from nine gastrointestinal stromal tumours, 14 synovial sarcomas, seven myxoid liposarcomas, 22 Ewing sarcomas and three Ewing-like small round cell tumours at a large sequencing depth to detect also mutations that are subclonal or occur at low allele frequencies. We found nine mutations in eight different potential driver genes, some of which are potentially actionable by currently existing targeted therapies. Even though no recurrent mutations in driver genes were found in the different sarcoma groups, we show that targeted NGS-based sequencing is clearly feasible in a diagnostic setting with very limited amounts of paraffin embedded tissue and may provide novel insights into mesenchymal cell signalling and potentially druggable targets. Interestingly, we also identify five non-synonymous sequence variants in 4 established cancer driver genes in DNA from normal tissue from sarcoma patients that may possibly predispose or contribute to neoplastic development.
Collapse
Affiliation(s)
- Carola Andersson
- Department of Clinical Pathology and Genetics, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
| | - Henrik Fagman
- Department of Clinical Pathology and Genetics, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
| | - Magnus Hansson
- Department of Clinical Pathology and Genetics, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
| | - Fredrik Enlund
- Department of Clinical Pathology and Genetics, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden.
| |
Collapse
|
34
|
Cornejo KM, Hutchinson L, Cyr MS, Nose V, McLaughlin PJ, Iafrate AJ, Sadow PM. MYC Analysis by Fluorescent In Situ Hybridization and Immunohistochemistry in Primary Adrenal Angiosarcoma (PAA): a Series of Four Cases. Endocr Pathol 2015. [PMID: 26223194 DOI: 10.1007/s12022-015-9385-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Primary adrenal angiosarcomas (PAA) are rare with 36 cases reported in the English literature. MYC protein expression and gene amplification have been detected in secondary angiosarcoma (AS), and a subset of primary AS. The aim of this study was to report the clinicopathologic features of PAA and examine these tumors for MYC amplification and protein expression in a small series of four cases (resection, n = 4). Three had available material for ancillary studies and were investigated for MYC gene abnormalities and protein expression using fluorescent in situ hybridization (FISH) and immunohistochemistry (IHC), respectively. Tumors occurred in three females and one male with a mean age of 69 (53-75) years. The sizes ranged from 8.5 to 15 (mean 11.5) cm and were epithelioid in morphology. All tumors had prominent necrosis, and the mitotic count ranged from 4 to 41/10 high-power fields (HPFs) (mean 20/10 HPFs, ×400). Immunohistochemically, the tumor cells were positive for CD31 in 4/4 cases, CD34 in 1/4 cases, and cytokeratin in 4/4 cases. The mean follow-up period was 10.8 (3-19) months, of which three patients died of disease with distant metastases, and one patient was alive with disease. MYC nuclear staining was identified in the three cases tested. Two cases showed polysomy of chromosome 8 without MYC amplification or rearrangement. Two MYC-positive cases by IHC demonstrated copy number gain in chromosome 8, and one MYC-positive case was not associated with a chromosome 8/MYC gene abnormality. In the context of new targeted therapies, MYC positivity in PAA may be clinically valuable in treating patients with these aggressive neoplasms.
Collapse
Affiliation(s)
- Kristine M Cornejo
- Department of Pathology, UMass Memorial Healthcare-University of Massachusetts Medical School, One Innovation Drive, Biotech 3, Worcester, MA, 01605, USA.
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Lloyd Hutchinson
- Department of Pathology, UMass Memorial Healthcare-University of Massachusetts Medical School, One Innovation Drive, Biotech 3, Worcester, MA, 01605, USA
| | - Maryann St Cyr
- Department of Pathology, UMass Memorial Healthcare-University of Massachusetts Medical School, One Innovation Drive, Biotech 3, Worcester, MA, 01605, USA
| | - Vania Nose
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Patrick J McLaughlin
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - A John Iafrate
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Peter M Sadow
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
35
|
Brenca M, Maestro R. Massive parallel sequencing in sarcoma pathobiology: state of the art and perspectives. Expert Rev Anticancer Ther 2015; 15:1473-88. [PMID: 26536249 DOI: 10.1586/14737140.2015.1108192] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Sarcomas are an aggressive and highly heterogeneous group of mesenchymal malignancies with different morphologies and clinical behavior. Current therapeutic strategies remain unsatisfactory. Cytogenetic and molecular characterization of these tumors is resulting in the breakdown of the classical histopathological categories into molecular subgroups that better define sarcoma pathobiology and pave the way to more precise diagnostic criteria and novel therapeutic opportunities. The purpose of this short review is to summarize the state-of-the-art on the exploitation of massive parallel sequencing technologies, also known as next generation sequencing, in the elucidation of sarcoma pathobiology and to discuss how these applications may impact on diagnosis, prognosis and therapy of these tumors.
Collapse
Affiliation(s)
- Monica Brenca
- a Experimental Oncology 1 , CRO Aviano National Cancer Institute , Aviano , PN 33081 , Italy
| | - Roberta Maestro
- a Experimental Oncology 1 , CRO Aviano National Cancer Institute , Aviano , PN 33081 , Italy
| |
Collapse
|
36
|
Brany D, Dvorska D, Nachajova M, Slavik P, Burjanivova T. Malignant tumors of the uterine corpus: molecular background of their origin. Tumour Biol 2015; 36:6615-21. [DOI: 10.1007/s13277-015-3824-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 07/20/2015] [Indexed: 12/21/2022] Open
|
37
|
Combined Targeted DNA Sequencing in Non-Small Cell Lung Cancer (NSCLC) Using UNCseq and NGScopy, and RNA Sequencing Using UNCqeR for the Detection of Genetic Aberrations in NSCLC. PLoS One 2015; 10:e0129280. [PMID: 26076459 PMCID: PMC4468211 DOI: 10.1371/journal.pone.0129280] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 05/06/2015] [Indexed: 01/21/2023] Open
Abstract
The recent FDA approval of the MiSeqDx platform provides a unique opportunity to develop targeted next generation sequencing (NGS) panels for human disease, including cancer. We have developed a scalable, targeted panel-based assay termed UNCseq, which involves a NGS panel of over 200 cancer-associated genes and a standardized downstream bioinformatics pipeline for detection of single nucleotide variations (SNV) as well as small insertions and deletions (indel). In addition, we developed a novel algorithm, NGScopy, designed for samples with sparse sequencing coverage to detect large-scale copy number variations (CNV), similar to human SNP Array 6.0 as well as small-scale intragenic CNV. Overall, we applied this assay to 100 snap-frozen lung cancer specimens lacking same-patient germline DNA (07–0120 tissue cohort) and validated our results against Sanger sequencing, SNP Array, and our recently published integrated DNA-seq/RNA-seq assay, UNCqeR, where RNA-seq of same-patient tumor specimens confirmed SNV detected by DNA-seq, if RNA-seq coverage depth was adequate. In addition, we applied the UNCseq assay on an independent lung cancer tumor tissue collection with available same-patient germline DNA (11–1115 tissue cohort) and confirmed mutations using assays performed in a CLIA-certified laboratory. We conclude that UNCseq can identify SNV, indel, and CNV in tumor specimens lacking germline DNA in a cost-efficient fashion.
Collapse
|
38
|
|
39
|
Sborov D, Chen JL. Targeted therapy in sarcomas other than GIST tumors. J Surg Oncol 2015; 111:632-40. [PMID: 25330750 PMCID: PMC4436975 DOI: 10.1002/jso.23802] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Accepted: 08/13/2014] [Indexed: 01/01/2023]
Abstract
Non-GIST soft tissue sarcomas are a heterogeneous grouping of mesenchymal tumors that comprise less than 1% of adult malignancies. Treatment continues to be based on cytotoxic chemotherapy regimens. However, characterization of the molecular pathway deregulations that drive these tumors has led to the emergence of more customized treatment options. In this review, we focus on the multitude of molecular inhibitors targeting angiogenesis and cell cycle pathways being tested in clinical trials.
Collapse
Affiliation(s)
- Douglas Sborov
- Hematology and Oncology Fellow, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - James L Chen
- Assistant Professor, Departments of Biomedical Informatics and Internal Medicine (Division of Medical Oncology), The Ohio State University, Columbus, OH, USA
| |
Collapse
|
40
|
Smith SM, Coleman J, Bridge JA, Iwenofu OH. Molecular diagnostics in soft tissue sarcomas and gastrointestinal stromal tumors. J Surg Oncol 2015; 111:520-31. [PMID: 25772665 DOI: 10.1002/jso.23882] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 12/11/2014] [Indexed: 12/17/2022]
Abstract
Soft tissue sarcomas are rare malignant heterogenous tumors of mesenchymal origin with over fifty subtypes. The use of hematoxylin and eosin stained sections (and immunohistochemistry) in the morphologic assessment of these tumors has been the bane of clinical diagnosis until recently. The last decade has witnessed considerable progress in the understanding and application of molecular techniques in refining the current understanding of soft tissue sarcomas and gastrointestinal stromal tumors beyond the limits of traditional approaches. Indeed, the identification of reciprocal chromosomal translocations and fusion genes in some subsets of sarcomas with potential implications in the pathogenesis, diagnosis and treatment has been revolutionary. The era of molecular targeted therapy presents a platform that continues to drive biomarker discovery and personalized medicine in soft tissue sarcomas and gastrointestinal stromal tumors. In this review, we highlight how the different molecular techniques have enhanced the diagnosis of these tumors with prognostic and therapeutic implications.
Collapse
Affiliation(s)
- Stephen M Smith
- Department of Pathology & Laboratory Medicine, Wexner Medical Center at The Ohio State University, Columbus, Ohio
| | | | | | | |
Collapse
|
41
|
Quintas V, Prada-López I, Donos N, Suárez-Quintanilla D, Tomás I. Antiplaque effect of essential oils and 0.2% chlorhexidine on an in situ model of oral biofilm growth: a randomised clinical trial. PLoS One 2015; 10:e0117177. [PMID: 25689859 PMCID: PMC4331278 DOI: 10.1371/journal.pone.0117177] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE To evaluate the in situ antiplaque effect after 4 days of using of 2 commercial antimicrobial agents in short term on undisturbed plaque-like biofilm. TRIAL DESIGN AND PARTICIPANTS An observer-masked, crossover randomised clinical trial on 15 oral and systemically healthy volunteers between 20-30 years who were randomly and sequentially allocated in the same group which performed 3 interventions in different randomised sequences. INTERVENTION The participants wore an appliance in 3 different rinsing periods doing mouthwashes twice a day (1/0/1) with essential oils, 0.2% chlorhexidine or sterile water (negative control). At the end of each 4-day mouthwash period, samples were removed from the appliance. Posteriorly, after bacterial vital staining, samples were analysed using a Confocal Laser Scanning Microscope. MAIN OUTCOME MEASURES Bacterial vitality, thickness and covering grade by the biofilm after 4 days of applying each of the mouthwashes. RESULTS The essential oils and the 0.2% chlorhexidine were significantly more effective than the sterile water at reducing bacterial vitality, thickness and covering grade by the biofilm. No significant differences were found between the 0.2% chlorhexidine and the essential oils at reducing the bacterial vitality (13.2% vs. 14.7%). However, the 0.2% chlorhexidine showed more reduction than the essential oils in thickness (6.5 μm vs. 10.0 μm; p<0.05) and covering grade by the biofilm (20.0% vs. 54.3%; p<0.001). CONCLUSION The essential oils and 0.2% chlorhexidine showed a high antiplaque effect. Although the 0.2% chlorhexidine showed better results with regard to reducing the thickness and covering grade by the biofilm, both antiseptics showed a high and similar antibacterial activity. CLINICAL RELEVANCE Daily essential oils or 0.2% chlorhexidine mouthwashes are effective when reducing dental plaque formation in the short term. Although 0.2% chlorhexidine continues to be the "gold standard" in terms of antiplaque effect, essential oils could be considered a reliable alternative. TRIAL REGISTRATION ClinicalTrials.gov NCT02124655.
Collapse
Affiliation(s)
- Víctor Quintas
- Oral Sciences Research Group, School of Medicine and Dentistry, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Isabel Prada-López
- Oral Sciences Research Group, School of Medicine and Dentistry, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Nikolaos Donos
- Periodontology Unit, UCL Eastman Dental Institute, London, United Kingdom
| | - David Suárez-Quintanilla
- Oral Sciences Research Group, School of Medicine and Dentistry, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Inmaculada Tomás
- Oral Sciences Research Group, School of Medicine and Dentistry, University of Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|